首页 > 最新文献

CNS & neurological disorders drug targets最新文献

英文 中文
Parkinson's Disease: Unravelling the Medicinal Perspectives and Recent Developments of Heterocyclic Monoamine Oxidase-B Inhibitors. 帕金森病:揭示杂环单胺氧化酶-B 抑制剂的药用前景和最新发展。
Pub Date : 2024-11-04 DOI: 10.2174/0118715273340983241018095529
Neha Rana, Parul Grover

Parkinson's disease is a neurodegenerative condition characterized by slow movement (bradykinesia), tremors, and muscle stiffness. These symptoms occur due to the degeneration of dopamine- producing neurons in the substantia nigra region of the brain, leading to reduced dopamine levels. The development of Parkinson's Disease (PD) involves a combination of genetic and environmental factors. PD is associated with abnormal regulation of the monoamine oxidase (MAO) enzyme. Monoamine oxidase inhibitors (MAOIs) are an important class of drugs used to treat PD and other neurological disorders. In the early stages of PD, monotherapy with MAO-B inhibitors has been shown to be both safe and effective. These inhibitors are also commonly used as adjuncts in long-term disease management, as they can improve both motor and non-motor symptoms, reduce "OFF" periods, and potentially slow disease progression. However, current MAO-B inhibitors come with side effects like dizziness, nausea, vomiting, light-headedness, and fainting. Therefore, accelerating the development of new MAO-B inhibitors with fewer side effects is crucial. This review explores natural compounds that may inhibit monoamine oxidase B (MAO-B), focusing on key findings from the past seven years. It highlights the most effective heterocyclic compounds against MAO-B, including thiazolyl hydrazone, pyridoxine-resveratrol, pyridazine, isoxazole, oxadiazole, benzothiazole, benzoxazole, coumarin, caffeine, pyrazoline, piperazine, piperidine, pyrrolidine, and morpholine derivatives. The review covers in vitro, in silico, and in vivo data, along with the structure- activity relationship of these compounds. These findings offer valuable insights for the development of more effective MAO-B inhibitors and advancements in Parkinson's disease research.

帕金森病是一种神经退行性疾病,以运动缓慢(运动迟缓)、震颤和肌肉僵硬为特征。出现这些症状的原因是大脑黑质区域产生多巴胺的神经元发生变性,导致多巴胺水平降低。帕金森病(PD)的发病涉及遗传和环境因素的综合作用。帕金森病与单胺氧化酶(MAO)调节异常有关。单胺氧化酶抑制剂(MAOIs)是治疗帕金森病和其他神经系统疾病的一类重要药物。在帕金森氏症的早期阶段,使用 MAO-B 抑制剂进行单药治疗已被证明是安全有效的。这些抑制剂还常用于长期疾病管理的辅助治疗,因为它们可以改善运动和非运动症状,减少 "关机 "期,并有可能减缓疾病的进展。然而,目前的 MAO-B 抑制剂有头晕、恶心、呕吐、头重脚轻和昏厥等副作用。因此,加快开发副作用较少的新型 MAO-B 抑制剂至关重要。本综述探讨了可抑制单胺氧化酶 B(MAO-B)的天然化合物,重点关注过去七年的主要研究成果。它重点介绍了对 MAO-B 最有效的杂环化合物,包括噻唑腙、吡哆醇-白藜芦醇、哒嗪、异噁唑、噁二唑、苯并噻唑、苯并噁唑、香豆素、咖啡碱、吡唑啉、哌嗪、哌啶、吡咯烷和吗啉衍生物。综述涵盖了体外、硅学和体内数据,以及这些化合物的结构-活性关系。这些发现为开发更有效的 MAO-B 抑制剂和推进帕金森病研究提供了宝贵的见解。
{"title":"Parkinson's Disease: Unravelling the Medicinal Perspectives and Recent Developments of Heterocyclic Monoamine Oxidase-B Inhibitors.","authors":"Neha Rana, Parul Grover","doi":"10.2174/0118715273340983241018095529","DOIUrl":"https://doi.org/10.2174/0118715273340983241018095529","url":null,"abstract":"<p><p>Parkinson's disease is a neurodegenerative condition characterized by slow movement (bradykinesia), tremors, and muscle stiffness. These symptoms occur due to the degeneration of dopamine- producing neurons in the substantia nigra region of the brain, leading to reduced dopamine levels. The development of Parkinson's Disease (PD) involves a combination of genetic and environmental factors. PD is associated with abnormal regulation of the monoamine oxidase (MAO) enzyme. Monoamine oxidase inhibitors (MAOIs) are an important class of drugs used to treat PD and other neurological disorders. In the early stages of PD, monotherapy with MAO-B inhibitors has been shown to be both safe and effective. These inhibitors are also commonly used as adjuncts in long-term disease management, as they can improve both motor and non-motor symptoms, reduce \"OFF\" periods, and potentially slow disease progression. However, current MAO-B inhibitors come with side effects like dizziness, nausea, vomiting, light-headedness, and fainting. Therefore, accelerating the development of new MAO-B inhibitors with fewer side effects is crucial. This review explores natural compounds that may inhibit monoamine oxidase B (MAO-B), focusing on key findings from the past seven years. It highlights the most effective heterocyclic compounds against MAO-B, including thiazolyl hydrazone, pyridoxine-resveratrol, pyridazine, isoxazole, oxadiazole, benzothiazole, benzoxazole, coumarin, caffeine, pyrazoline, piperazine, piperidine, pyrrolidine, and morpholine derivatives. The review covers in vitro, in silico, and in vivo data, along with the structure- activity relationship of these compounds. These findings offer valuable insights for the development of more effective MAO-B inhibitors and advancements in Parkinson's disease research.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142585260","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhanced Neuroprotection in Experiment Multiple Sclerosis through Combined Rosiglitazone and Probiotic-Loaded Solid Lipid Nanoparticles: Modulation of Cellular Signaling Pathways. 联合使用罗格列酮和益生菌载体固体脂质纳米颗粒增强实验性多发性硬化症的神经保护:细胞信号通路的调节。
Pub Date : 2024-11-01 DOI: 10.2174/0118715273336107241015100912
Nitish Kumar, Nidhi Tyagi, Sidharth Mehan, Alok Pratap Singh

Background: Multiple sclerosis (MS) is a persistent autoimmune condition characterized by inflammation and neurodegeneration. The current efficacy of treatments is limited, which has generated interest in developing neuroprotective strategies. Solid lipid nanoparticles (SLNs) and probiotics are potential drug delivery vehicles for targeting the CNS (Central nervous system), regulating immune responses, and supporting neuroprotection in neurological conditions.

Methods: The study investigates how SLNs containing RSG (rosiglitazone) and probiotics can protect the nervous system in cases of MS. We administered toxin EtBr (Ethidium bromide) from day 1 to day 7, later followed by the treatment from day 8 to day 35. During this time interval, various behavioural parameters have been performed. Further, after 35th day, blood plasma of animals was collected to study complete CBC profiling and animals were sacrificed. Then, biochemical and molecular studies, gross morphology of brain sectioning, histopathological evaluation and estimation of fatty acid content in fecal matter were performed.

Results: RSG shows neuroprotective effects by blocking the STAT-3 and mTOR signaling pathways and increasing the production of PPAR-gamma. GW9662, a PPAR-gamma antagonist given at a dose of 2 mg/kg (i.p), was utilized to evaluate the role of PPAR-gamma and to compare the efficacy of RSG and probiotic-loaded SLNs in potentially providing neuroprotection. The relationship between RSG and the STAT-3, mTOR, and PPAR-gamma pathways in MS was confirmed and validated using in-silico analysis. RSG and probiotic-loaded SLNs modulate the complete blood profiling of rats and improve the symptoms of MS. We assessed the diagnostic capabilities of different biological samples such as cerebrospinal fluid, blood plasma, and brain homogenates (specifically from the hippocampus, striatum, cortex, and midbrain) to analyze neurochemical changes linked to neurobehavioral changes in the progression of MS.

Conclusion: The study showed that combining RSG and probiotics in an experimental medication form improved symptoms of MS more effectively than using RSG alone. This improvement is likely due to changes in STAT-3, mTOR, and PPAR-gamma signaling pathways.

背景:多发性硬化症(MS)是一种以炎症和神经变性为特征的顽固性自身免疫疾病。目前的治疗效果有限,这引起了人们对开发神经保护策略的兴趣。固体脂质纳米颗粒(SLNs)和益生菌是潜在的药物输送载体,可用于靶向中枢神经系统(CNS)、调节免疫反应和支持神经系统疾病的神经保护:本研究探讨了含有 RSG(罗格列酮)和益生菌的 SLNs 如何保护多发性硬化症患者的神经系统。我们从第1天到第7天施用毒素EtBr(溴化乙锭),随后从第8天到第35天进行治疗。在此期间,我们检测了各种行为参数。此外,在第 35 天后,收集动物血浆以研究完整的全血细胞计数,并将动物处死。然后,进行生化和分子研究、脑切片大体形态学研究、组织病理学评估和粪便中脂肪酸含量的估计:结果:RSG通过阻断STAT-3和mTOR信号通路以及增加PPAR-gamma的产生而显示出神经保护作用。GW9662是一种PPAR-γ拮抗剂,给药剂量为2毫克/千克(静脉注射),用于评估PPAR-γ的作用,并比较RSG和装载益生菌的SLNs在提供潜在神经保护方面的功效。通过使用in-silico分析,确认并验证了RSG与多发性硬化症中STAT-3、mTOR和PPAR-gamma通路之间的关系。RSG 和装载益生菌的 SLN 调节了大鼠的全血谱分析,改善了多发性硬化症的症状。我们评估了脑脊液、血浆和脑匀浆(特别是海马、纹状体、皮层和中脑)等不同生物样本的诊断能力,以分析多发性硬化症进展过程中与神经行为变化相关的神经化学变化:研究表明,与单独使用 RSG 相比,将 RSG 和益生菌以实验性药物形式结合使用能更有效地改善多发性硬化症的症状。这种改善可能是由于 STAT-3、mTOR 和 PPAR-gamma 信号通路发生了变化。
{"title":"Enhanced Neuroprotection in Experiment Multiple Sclerosis through Combined Rosiglitazone and Probiotic-Loaded Solid Lipid Nanoparticles: Modulation of Cellular Signaling Pathways.","authors":"Nitish Kumar, Nidhi Tyagi, Sidharth Mehan, Alok Pratap Singh","doi":"10.2174/0118715273336107241015100912","DOIUrl":"https://doi.org/10.2174/0118715273336107241015100912","url":null,"abstract":"<p><strong>Background: </strong>Multiple sclerosis (MS) is a persistent autoimmune condition characterized by inflammation and neurodegeneration. The current efficacy of treatments is limited, which has generated interest in developing neuroprotective strategies. Solid lipid nanoparticles (SLNs) and probiotics are potential drug delivery vehicles for targeting the CNS (Central nervous system), regulating immune responses, and supporting neuroprotection in neurological conditions.</p><p><strong>Methods: </strong>The study investigates how SLNs containing RSG (rosiglitazone) and probiotics can protect the nervous system in cases of MS. We administered toxin EtBr (Ethidium bromide) from day 1 to day 7, later followed by the treatment from day 8 to day 35. During this time interval, various behavioural parameters have been performed. Further, after 35th day, blood plasma of animals was collected to study complete CBC profiling and animals were sacrificed. Then, biochemical and molecular studies, gross morphology of brain sectioning, histopathological evaluation and estimation of fatty acid content in fecal matter were performed.</p><p><strong>Results: </strong>RSG shows neuroprotective effects by blocking the STAT-3 and mTOR signaling pathways and increasing the production of PPAR-gamma. GW9662, a PPAR-gamma antagonist given at a dose of 2 mg/kg (i.p), was utilized to evaluate the role of PPAR-gamma and to compare the efficacy of RSG and probiotic-loaded SLNs in potentially providing neuroprotection. The relationship between RSG and the STAT-3, mTOR, and PPAR-gamma pathways in MS was confirmed and validated using in-silico analysis. RSG and probiotic-loaded SLNs modulate the complete blood profiling of rats and improve the symptoms of MS. We assessed the diagnostic capabilities of different biological samples such as cerebrospinal fluid, blood plasma, and brain homogenates (specifically from the hippocampus, striatum, cortex, and midbrain) to analyze neurochemical changes linked to neurobehavioral changes in the progression of MS.</p><p><strong>Conclusion: </strong>The study showed that combining RSG and probiotics in an experimental medication form improved symptoms of MS more effectively than using RSG alone. This improvement is likely due to changes in STAT-3, mTOR, and PPAR-gamma signaling pathways.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142570703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Parkinson's Disease: A Progressive Neurodegenerative Disorder and Structure-Activity Relationship of MAO Inhibitor Scaffolds as an Important Therapeutic Regimen. 帕金森病:帕金森病:一种进行性神经退行性疾病和作为重要治疗方案的 MAO 抑制剂支架的结构-活性关系。
Pub Date : 2024-10-30 DOI: 10.2174/0118715273324300241010054029
Salauddin, Syed Amir Azam Zaidi, Mohammed Ubaid, Saniya Shamim, Mohd Javed Naim, Suruchi Khanna, Ozair Alam

Parkinson's disease is considered an advancing neurodegenerative disorder with unknown causes, and its association with some risk factors, including aging, family history, and exposure to chemicals, makes it the second most common occurring neurodegenerative disorder throughout the world with increasing prevalence. Parkinson's disease is associated with slow movement, rigidity, tremors, imbalance, depression, anxiety, cognitive impairment, orthostasis, hyperhidrosis, sleep disorders, pain, and sensory disturbances. In recent decades, there has been a rise in research on the development of effective and potential therapies for the treatment of Parkinson's disease. An important target for neuroprotection is Monoamine Oxidases (MAO), which hydrolyze neurotransmitters like dopamine and produce very reactive free radicals as a by-product. Aging and neurodegenerative illnesses cause overexpression in the brain, which exacerbates neuronal loss. The treatment of Parkinson's disease with MAO inhibitors has shown promising outcomes. Herein, we reported characteristic features of Parkinson's disease, various treatment strategies, and the SAR of potential drugs that can be explored further as lead for the development of newer molecules with improved pharmacological profiles.

帕金森病被认为是一种病因不明的渐进性神经退行性疾病,它与一些风险因素(包括衰老、家族史和接触化学物质)有关,因此是全球第二大最常见的神经退行性疾病,而且发病率越来越高。帕金森病与运动迟缓、僵直、震颤、失衡、抑郁、焦虑、认知障碍、正位、多汗、睡眠障碍、疼痛和感觉障碍有关。近几十年来,有关开发治疗帕金森病的有效和潜在疗法的研究不断增加。神经保护的一个重要靶点是单胺氧化酶(MAO),它能水解多巴胺等神经递质,并产生活性很强的自由基作为副产品。衰老和神经退行性疾病会导致单胺氧化酶在大脑中过度表达,从而加剧神经元的损失。用 MAO 抑制剂治疗帕金森病已显示出良好的疗效。在此,我们报告了帕金森病的特征、各种治疗策略以及潜在药物的 SAR,这些药物可作为开发药理特征更佳的新分子的先导药物进行进一步探索。
{"title":"Parkinson's Disease: A Progressive Neurodegenerative Disorder and Structure-Activity Relationship of MAO Inhibitor Scaffolds as an Important Therapeutic Regimen.","authors":"Salauddin, Syed Amir Azam Zaidi, Mohammed Ubaid, Saniya Shamim, Mohd Javed Naim, Suruchi Khanna, Ozair Alam","doi":"10.2174/0118715273324300241010054029","DOIUrl":"https://doi.org/10.2174/0118715273324300241010054029","url":null,"abstract":"<p><p>Parkinson's disease is considered an advancing neurodegenerative disorder with unknown causes, and its association with some risk factors, including aging, family history, and exposure to chemicals, makes it the second most common occurring neurodegenerative disorder throughout the world with increasing prevalence. Parkinson's disease is associated with slow movement, rigidity, tremors, imbalance, depression, anxiety, cognitive impairment, orthostasis, hyperhidrosis, sleep disorders, pain, and sensory disturbances. In recent decades, there has been a rise in research on the development of effective and potential therapies for the treatment of Parkinson's disease. An important target for neuroprotection is Monoamine Oxidases (MAO), which hydrolyze neurotransmitters like dopamine and produce very reactive free radicals as a by-product. Aging and neurodegenerative illnesses cause overexpression in the brain, which exacerbates neuronal loss. The treatment of Parkinson's disease with MAO inhibitors has shown promising outcomes. Herein, we reported characteristic features of Parkinson's disease, various treatment strategies, and the SAR of potential drugs that can be explored further as lead for the development of newer molecules with improved pharmacological profiles.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142559739","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring Therapeutic Strategies: The Relationship between Metabolic Disorders and FOXO Signalling in Alzheimer's Disease. 探索治疗策略:阿尔茨海默病中代谢紊乱与 FOXO 信号之间的关系。
Pub Date : 2024-10-28 DOI: 10.2174/0118715273321002240919102841
Parneet Kaur, Heena Khan, Amarjot Kaur Grewal, Kamal Dua, Sachin Kumar Singh, Gaurav Gupta, Thakur Gurjeet Singh

Alzheimer's disease is an ailment that is linked with the degeneration of the brain cells, and this illness is the main cause of dementia. Metabolic stress affects the activity of the brain in AD via FOXO signaling. The occurrence of AD will significantly surge as the world's population ages, along with lifestyle changes perceived in current decades, indicating a main contributor to such augmented prevalence. Similarly, metabolic disorders of current adulthood, such as obesity, stroke, and diabetes mellitus, have been observed as the risk-causing factors of AD. Environmental influences induce genetic mutations that result in the development of several diseases. Metabolic disorders develop when individuals are exposed to an environment where food is easily accessible and requires minimal energy expenditure. Obesity and diabetes are among the most significant worldwide health concerns. Obesity arises because of an imbalance between the amount of energy consumed and the amount of energy expended, which is caused by both behavioral and physiological factors. Obesity, insulin resistance syndrome, hypertension, and inflammation are factors that contribute to the worldwide risk of developing diabetes mellitus and neurodegenerative diseases. FOXO transcription factors are preserved molecules that play an important part in assorted biological progressions, precisely in aging as well as metabolism. Apoptosis, cell division and differentiation, oxidative stress, metabolism, and lifespan are among the physiological processes that the FOXO proteins are adept at controlling. In this review, we explored the correlation between signaling pathways and the cellular functions of FOXO proteins. We have also summarized the intricate role of FOXO in AD, with a focus on metabolic stress, and discussed the prospect of FOXO as a molecular link between AD and metabolic disorders.

阿尔茨海默病是一种与脑细胞变性有关的疾病,这种疾病是导致痴呆症的主要原因。代谢压力会通过 FOXO 信号转导影响老年痴呆症患者大脑的活动。随着世界人口的老龄化,以及当前几十年生活方式的改变,注意力缺失症的发病率将大幅上升,这表明注意力缺失症是导致发病率上升的主要原因。同样,肥胖、中风和糖尿病等成年期代谢紊乱也被认为是导致注意力缺失症的危险因素。环境影响会诱发基因突变,导致多种疾病的发生。当个体处于食物容易获取且只需极少能量消耗的环境中时,就会出现代谢紊乱。肥胖症和糖尿病是全球最重要的健康问题之一。肥胖症的产生是由于行为和生理因素造成的能量消耗和能量消耗之间的不平衡。肥胖、胰岛素抵抗综合征、高血压和炎症是导致全球糖尿病和神经退行性疾病发病风险的因素。FOXO 转录因子是一种保留下来的分子,在各种生物进程中发挥着重要作用,尤其是在衰老和新陈代谢方面。细胞凋亡、细胞分裂和分化、氧化应激、新陈代谢和寿命都是 FOXO 蛋白擅长控制的生理过程。在这篇综述中,我们探讨了信号通路与 FOXO 蛋白的细胞功能之间的相关性。我们还总结了 FOXO 在注意力缺失症中的复杂作用,重点是代谢压力,并讨论了 FOXO 作为注意力缺失症和代谢紊乱之间的分子联系的前景。
{"title":"Exploring Therapeutic Strategies: The Relationship between Metabolic Disorders and FOXO Signalling in Alzheimer's Disease.","authors":"Parneet Kaur, Heena Khan, Amarjot Kaur Grewal, Kamal Dua, Sachin Kumar Singh, Gaurav Gupta, Thakur Gurjeet Singh","doi":"10.2174/0118715273321002240919102841","DOIUrl":"https://doi.org/10.2174/0118715273321002240919102841","url":null,"abstract":"<p><p>Alzheimer's disease is an ailment that is linked with the degeneration of the brain cells, and this illness is the main cause of dementia. Metabolic stress affects the activity of the brain in AD via FOXO signaling. The occurrence of AD will significantly surge as the world's population ages, along with lifestyle changes perceived in current decades, indicating a main contributor to such augmented prevalence. Similarly, metabolic disorders of current adulthood, such as obesity, stroke, and diabetes mellitus, have been observed as the risk-causing factors of AD. Environmental influences induce genetic mutations that result in the development of several diseases. Metabolic disorders develop when individuals are exposed to an environment where food is easily accessible and requires minimal energy expenditure. Obesity and diabetes are among the most significant worldwide health concerns. Obesity arises because of an imbalance between the amount of energy consumed and the amount of energy expended, which is caused by both behavioral and physiological factors. Obesity, insulin resistance syndrome, hypertension, and inflammation are factors that contribute to the worldwide risk of developing diabetes mellitus and neurodegenerative diseases. FOXO transcription factors are preserved molecules that play an important part in assorted biological progressions, precisely in aging as well as metabolism. Apoptosis, cell division and differentiation, oxidative stress, metabolism, and lifespan are among the physiological processes that the FOXO proteins are adept at controlling. In this review, we explored the correlation between signaling pathways and the cellular functions of FOXO proteins. We have also summarized the intricate role of FOXO in AD, with a focus on metabolic stress, and discussed the prospect of FOXO as a molecular link between AD and metabolic disorders.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549481","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Proposed Hypothesis of TWEAK/Fn14 Receptor Modulation in Autism Spectrum Disorder. 自闭症谱系障碍中 TWEAK/Fn14 受体调节的假说。
Pub Date : 2024-10-28 DOI: 10.2174/0118715273330549241015073953
Heena Khan, Vivek Rihal, Amarjot Kaur, Thakur Gurjeet Singh

Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with a complex, multiple etiology that is marked by impaired social interaction, communication, and repetitive behaviour. There is presently no pharmaceutical treatment for the core symptoms of ASD, even though the prevalence of ASD is increasing worldwide. Treatment of autism spectrum disorder involves the interaction of numerous signalling pathways, such as the Wnt/beta-catenin pathway, probiotics and kynurenine pathway, PPAR pathway, PI3K-AKT-mTOR pathway, Hedgehog signaling pathway, etc. The scientific literature has revealed TWEAK/Fn14 to not be explored in the autism spectrum disorder. In vitro and in vivo, TWEAK can control a wide range of cellular responses. Recent research has revealed that TWEAK and Fn14 are expressed in the Central Nervous System (CNS) and upregulated in perivascular endothelial cells, astrocytes, neurons, and microglia in response to various stimuli, including cerebral ischemia. This upregulation is followed by cell death and an increase in Blood-brain Barrier (BBB) permeability. The study has revealed that Aurintricarboxylic Acid (ATA) acts as an agent that suppresses TWEAK/Fn14 signaling. Similarly, from the discussion, it has been emphasized that the proposed molecular TWEAK/Fn14 signalling pathway can be considered as a therapeutic approach in the management of autism spectrum disorder.

自闭症谱系障碍(ASD)是一种神经发育障碍,病因复杂多样,主要表现为社交、沟通和重复行为障碍。尽管自闭症的发病率在全球范围内不断上升,但目前还没有针对自闭症核心症状的药物治疗方法。自闭症谱系障碍的治疗涉及众多信号通路的相互作用,如 Wnt/beta-catenin 通路、益生菌和犬尿氨酸通路、PPAR 通路、PI3K-AKT-mTOR 通路、刺猬信号通路等。科学文献显示,TWEAK/Fn14 在自闭症谱系障碍中并没有被发现。在体外和体内,TWEAK 可控制多种细胞反应。最近的研究发现,TWEAK 和 Fn14 在中枢神经系统(CNS)中表达,并在血管周围内皮细胞、星形胶质细胞、神经元和小胶质细胞中上调,以应对包括脑缺血在内的各种刺激。这种上调会导致细胞死亡和血脑屏障(BBB)通透性增加。研究发现,三羧酸(ATA)是一种抑制 TWEAK/Fn14 信号传导的物质。同样,讨论还强调,所提出的 TWEAK/Fn14 信号分子通路可被视为治疗自闭症谱系障碍的一种方法。
{"title":"Proposed Hypothesis of TWEAK/Fn14 Receptor Modulation in Autism Spectrum Disorder.","authors":"Heena Khan, Vivek Rihal, Amarjot Kaur, Thakur Gurjeet Singh","doi":"10.2174/0118715273330549241015073953","DOIUrl":"https://doi.org/10.2174/0118715273330549241015073953","url":null,"abstract":"<p><p>Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with a complex, multiple etiology that is marked by impaired social interaction, communication, and repetitive behaviour. There is presently no pharmaceutical treatment for the core symptoms of ASD, even though the prevalence of ASD is increasing worldwide. Treatment of autism spectrum disorder involves the interaction of numerous signalling pathways, such as the Wnt/beta-catenin pathway, probiotics and kynurenine pathway, PPAR pathway, PI3K-AKT-mTOR pathway, Hedgehog signaling pathway, etc. The scientific literature has revealed TWEAK/Fn14 to not be explored in the autism spectrum disorder. In vitro and in vivo, TWEAK can control a wide range of cellular responses. Recent research has revealed that TWEAK and Fn14 are expressed in the Central Nervous System (CNS) and upregulated in perivascular endothelial cells, astrocytes, neurons, and microglia in response to various stimuli, including cerebral ischemia. This upregulation is followed by cell death and an increase in Blood-brain Barrier (BBB) permeability. The study has revealed that Aurintricarboxylic Acid (ATA) acts as an agent that suppresses TWEAK/Fn14 signaling. Similarly, from the discussion, it has been emphasized that the proposed molecular TWEAK/Fn14 signalling pathway can be considered as a therapeutic approach in the management of autism spectrum disorder.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549483","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In Silico and ADMET Studies of Spiro-Quinazoline Compounds as Acetylcholine Esterase Inhibitors Against Alzheimer's Disease. 螺喹唑啉化合物作为乙酰胆碱酯酶抑制剂防治阿尔茨海默病的硅学和 ADMET 研究。
Pub Date : 2024-10-25 DOI: 10.2174/0118715273315412241009092249
Abdulelah Aljuaid, Osama Abdulaziz, Mamdouh Allahyani, Mazen Almehmadi, Abdullah Yahya Abdullah Alzahrani, Shivani Verma, Mohd Yusuf, Mohammad Asif

Background: Alzheimer's disease (AD) is a prevalent neurodegenerative condition characterized by progressive cognitive decline and memory impairment resulting from the degeneration and death of brain neurons. Acetylcholinesterase (AChE) inhibitors are used in primary pharmacotherapy for numerous neurodegenerative conditions, providing their capacity to modulate acetylcholine levels crucial for cognitive function. Recently, quinazoline derivatives have emerged as a compelling model for neurodegenerative disease treatment, showcasing promising pharmacological features. Their unique structural features and pharmacokinetic profiles have sparked interest in their potential efficacy and safety across diverse neurodegenerative disorders. The exposure of quinazoline derivatives as a potential therapeutic way underscores the imperative for continued research exploration. Their multifaceted mechanisms of action and ability to target various pathways implicated in neurodegeneration offer exciting prospects for developing novel, effective, and well-tolerated treatments. Further investigations into their pharmacological activities and precise therapeutic roles are essential to advance our understanding of neurodegenerative disease pathophysiology and promote the development of modern therapeutic strategies to address this critical medical challenge.

Methods: Quinazoline derivatives have gained eminent acetylcholinesterase (AChE) inhibitory activity. Their ability to effectively modulate AChE activity makes them promising candidates for treating neurological disorders, particularly Alzheimer's disease (AD). Their intricate molecular structures confer selectivity and affinity for AChE, offering potential for the development of novel therapeutic agents targeting cholinergic pathways. Hence, in this study, we designed, synthesized, and characterized a series of spiro[cycloalakane-1,2'-quinazoline derivatives (1-6) to assess their possible AChE inhibiting ability using docking into the active sites.

Results: The AChE inhibitory potential of spiro[cycloalkane-1,2'-quinazoline derivatives (1-6) was explored via docking studies of the AChE active site. The findings revealed significant inhibitory activity and highlighted the promising nature of these derivatives.

Conclusion: The synthesized spiro[cycloalkane-1,2'-quinazoline derivatives (1-6) exhibited their notable potential as AChE inhibitors. The observed significant inhibitory activity suggested that these derivatives warrant further exploration as candidates for developing therapeutic agents in AChE inhibitory pathways. This study emphasizes the relevance of quinazoline derivatives in searching for novel treatments for neurological disorders, particularly associated with cholinergic dysfunction, and they could be a useful alternative therapeutic agent.

背景:阿尔茨海默病(AD)是一种常见的神经退行性疾病,其特征是由于大脑神经元的变性和死亡而导致的进行性认知能力下降和记忆障碍。乙酰胆碱酯酶(AChE)抑制剂可调节对认知功能至关重要的乙酰胆碱水平,因此被用于多种神经退行性疾病的主要药物治疗。最近,喹唑啉衍生物作为一种引人注目的神经退行性疾病治疗模式出现了,并展示出良好的药理特性。它们独特的结构特征和药代动力学特征引发了人们对其在各种神经退行性疾病中潜在疗效和安全性的兴趣。喹唑啉衍生物作为一种潜在的治疗方法,强调了继续研究探索的必要性。喹唑啉衍生物具有多方面的作用机制,能够靶向与神经退行性病变有关的各种途径,为开发新型、有效、耐受性良好的治疗方法提供了令人振奋的前景。进一步研究喹唑啉衍生物的药理活性和确切的治疗作用,对于增进我们对神经退行性疾病病理生理学的了解和促进现代治疗策略的开发以应对这一严峻的医学挑战至关重要:喹唑啉衍生物具有显著的乙酰胆碱酯酶(AChE)抑制活性。喹唑啉衍生物能有效调节乙酰胆碱酯酶的活性,因此有望用于治疗神经系统疾病,尤其是阿尔茨海默病(AD)。它们错综复杂的分子结构赋予了对 AChE 的选择性和亲和性,为开发针对胆碱能通路的新型治疗药物提供了潜力。因此,在本研究中,我们设计、合成并表征了一系列螺[环卡拉烷-1,2'-喹唑啉衍生物(1-6),利用与活性位点的对接来评估它们可能的 AChE 抑制能力:结果:通过对 AChE 活性位点的对接研究,探讨了螺[环烷烃-1,2'-喹唑啉衍生物 (1-6) 的 AChE 抑制潜力。研究结果表明,这些衍生物具有明显的抑制活性,并凸显了其广阔的前景:结论:合成的螺[环烷-1,2'-喹唑啉衍生物(1-6)显示了其作为 AChE 抑制剂的显著潜力。观察到的明显抑制活性表明,这些衍生物值得进一步探索,以开发 AChE 抑制途径的候选治疗药物。这项研究强调了喹唑啉衍生物在寻找神经系统疾病(尤其是与胆碱能功能障碍有关的疾病)的新型治疗方法方面的相关性,它们可以成为一种有用的替代治疗剂。
{"title":"In Silico and ADMET Studies of Spiro-Quinazoline Compounds as Acetylcholine Esterase Inhibitors Against Alzheimer's Disease.","authors":"Abdulelah Aljuaid, Osama Abdulaziz, Mamdouh Allahyani, Mazen Almehmadi, Abdullah Yahya Abdullah Alzahrani, Shivani Verma, Mohd Yusuf, Mohammad Asif","doi":"10.2174/0118715273315412241009092249","DOIUrl":"https://doi.org/10.2174/0118715273315412241009092249","url":null,"abstract":"<p><strong>Background: </strong>Alzheimer's disease (AD) is a prevalent neurodegenerative condition characterized by progressive cognitive decline and memory impairment resulting from the degeneration and death of brain neurons. Acetylcholinesterase (AChE) inhibitors are used in primary pharmacotherapy for numerous neurodegenerative conditions, providing their capacity to modulate acetylcholine levels crucial for cognitive function. Recently, quinazoline derivatives have emerged as a compelling model for neurodegenerative disease treatment, showcasing promising pharmacological features. Their unique structural features and pharmacokinetic profiles have sparked interest in their potential efficacy and safety across diverse neurodegenerative disorders. The exposure of quinazoline derivatives as a potential therapeutic way underscores the imperative for continued research exploration. Their multifaceted mechanisms of action and ability to target various pathways implicated in neurodegeneration offer exciting prospects for developing novel, effective, and well-tolerated treatments. Further investigations into their pharmacological activities and precise therapeutic roles are essential to advance our understanding of neurodegenerative disease pathophysiology and promote the development of modern therapeutic strategies to address this critical medical challenge.</p><p><strong>Methods: </strong>Quinazoline derivatives have gained eminent acetylcholinesterase (AChE) inhibitory activity. Their ability to effectively modulate AChE activity makes them promising candidates for treating neurological disorders, particularly Alzheimer's disease (AD). Their intricate molecular structures confer selectivity and affinity for AChE, offering potential for the development of novel therapeutic agents targeting cholinergic pathways. Hence, in this study, we designed, synthesized, and characterized a series of spiro[cycloalakane-1,2'-quinazoline derivatives (1-6) to assess their possible AChE inhibiting ability using docking into the active sites.</p><p><strong>Results: </strong>The AChE inhibitory potential of spiro[cycloalkane-1,2'-quinazoline derivatives (1-6) was explored via docking studies of the AChE active site. The findings revealed significant inhibitory activity and highlighted the promising nature of these derivatives.</p><p><strong>Conclusion: </strong>The synthesized spiro[cycloalkane-1,2'-quinazoline derivatives (1-6) exhibited their notable potential as AChE inhibitors. The observed significant inhibitory activity suggested that these derivatives warrant further exploration as candidates for developing therapeutic agents in AChE inhibitory pathways. This study emphasizes the relevance of quinazoline derivatives in searching for novel treatments for neurological disorders, particularly associated with cholinergic dysfunction, and they could be a useful alternative therapeutic agent.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549482","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Vitamin D and Neurological Health: Unraveling Risk Factors, Disease Progression, and Treatment Potential. 维生素 D 与神经系统健康:揭示风险因素、疾病进展和治疗潜力。
Pub Date : 2024-10-22 DOI: 10.2174/0118715273330972241009092828
Abdullah Al Noman, Halima Afrosa, Imanul Kabir Lihu, Onusree Sarkar, Naimur Rahman Nabin, Monty Datta, Rashmi Pathak, Himanshu Sharma

This study explores the complex link between vitamin D and neurological illnesses, focusing on how vitamin D affects possible risk factors, therapeutic applications, and the trajectory of the disease. An epidemiological study has linked vitamin D insufficiency to several neurological conditions, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. It is hypothesized that immunomodulatory and anti-inflammatory properties of vitamin D contribute to its neuroprotective effects. Two major mechanisms in dementia include neuroinflammation and oxidative stress. Adequate levels of vitamin D have been shown in both animal models and human studies to enhance both clinical outcomes and the duration of illness in those who have it. Other ways that vitamin D contributes to its therapeutic potential include the production of neurotrophic factors, control over neurotransmitter synthesis, and preservation of the blood-brain barrier. Despite the encouraging outcomes, research is still being conducted to determine the optimal dosage and long-term benefits of vitamin D supplementation on brain function. In order to furnish precise directives and clarify the processes behind the neuroprotective impacts of vitamin D, future research must focus on large-scale randomized controlled studies. . This study highlights the significance of maintaining adequate levels of vitamin D as a modifiable risk factor for neurological disorders. Further study is also required to comprehend the possible medical benefits of this vitamin fully.

本研究探讨了维生素 D 与神经系统疾病之间的复杂联系,重点是维生素 D 如何影响可能的风险因素、治疗应用以及疾病的发展轨迹。一项流行病学研究表明,维生素 D 不足与帕金森病、阿尔茨海默病和多发性硬化症等几种神经系统疾病有关。据推测,维生素 D 的免疫调节和抗炎特性有助于其神经保护作用。痴呆症的两个主要机制包括神经炎症和氧化应激。动物模型和人体研究都表明,充足水平的维生素 D 可提高临床疗效,延长患者的病程。维生素 D 发挥治疗潜力的其他途径包括产生神经营养因子、控制神经递质合成和保护血脑屏障。尽管取得了令人鼓舞的成果,但目前仍在进行研究,以确定补充维生素 D 的最佳剂量和对大脑功能的长期益处。为了提供准确的指导并阐明维生素 D 对神经保护作用背后的过程,未来的研究必须侧重于大规模随机对照研究。.这项研究强调了保持足够的维生素 D 水平作为神经系统疾病可调节风险因素的重要性。要充分了解这种维生素可能带来的医疗益处,还需要进一步的研究。
{"title":"Vitamin D and Neurological Health: Unraveling Risk Factors, Disease Progression, and Treatment Potential.","authors":"Abdullah Al Noman, Halima Afrosa, Imanul Kabir Lihu, Onusree Sarkar, Naimur Rahman Nabin, Monty Datta, Rashmi Pathak, Himanshu Sharma","doi":"10.2174/0118715273330972241009092828","DOIUrl":"https://doi.org/10.2174/0118715273330972241009092828","url":null,"abstract":"<p><p>This study explores the complex link between vitamin D and neurological illnesses, focusing on how vitamin D affects possible risk factors, therapeutic applications, and the trajectory of the disease. An epidemiological study has linked vitamin D insufficiency to several neurological conditions, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. It is hypothesized that immunomodulatory and anti-inflammatory properties of vitamin D contribute to its neuroprotective effects. Two major mechanisms in dementia include neuroinflammation and oxidative stress. Adequate levels of vitamin D have been shown in both animal models and human studies to enhance both clinical outcomes and the duration of illness in those who have it. Other ways that vitamin D contributes to its therapeutic potential include the production of neurotrophic factors, control over neurotransmitter synthesis, and preservation of the blood-brain barrier. Despite the encouraging outcomes, research is still being conducted to determine the optimal dosage and long-term benefits of vitamin D supplementation on brain function. In order to furnish precise directives and clarify the processes behind the neuroprotective impacts of vitamin D, future research must focus on large-scale randomized controlled studies. . This study highlights the significance of maintaining adequate levels of vitamin D as a modifiable risk factor for neurological disorders. Further study is also required to comprehend the possible medical benefits of this vitamin fully.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142514738","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hypoxia-inducible Factor-1α Pathway in Cerebral Ischemia: From Molecular Mechanisms to Therapeutic Targets. 脑缺血中的缺氧诱导因子-1α通路:从分子机制到治疗靶点。
Pub Date : 2024-10-18 DOI: 10.2174/0118715273324551241008111827
Veerta Sharma, Thakur Gurjeet Singh

Introduction: Ischemic injury to the brain can result in a variety of life-threatening conditions, mortality, or varying degrees of disability. Hypoxia-inducible factor 1α (HIF 1α), an oxygen- sensitive transcription factor that controls the adaptive metabolic response to hypoxia, is a critical constituent of cerebral ischemia. It participates in numerous processes, such as metabolism, proliferation, and angiogenesis, and plays a major role in cerebral ischemia.

Methods: Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the pharmacological modulation of HIF-1α pathways for the treatment of cerebral ischemia.

Results: Various signalling pathways, such as Mitogen-activated protein kinase (MAPK), Janus kinase/ signal transducers and activators (JAK/STAT), Phosphoinositide-3-kinase (PI3-K), and cAMPresponse element binding protein (CREB) play a vital role in modulation of HIF-1α pathway, which helps in preventing the pathogenesis of cerebral ischemia.

Conclusion: The pharmacological modulation of the HIF-1α pathway via various molecular signalling pathways, such as PI3-K, MAPK, CREB, and JAK/STAT activators, offer a promising prospect for future interventions and treatment for cerebral ischemia.

导言:脑缺血损伤可导致多种危及生命的情况、死亡或不同程度的残疾。缺氧诱导因子 1α(HIF 1α)是一种对氧敏感的转录因子,可控制对缺氧的适应性代谢反应,是脑缺血的重要组成部分。它参与代谢、增殖和血管生成等多个过程,在脑缺血中发挥着重要作用:方法:通过使用 Scopus、PubMed、Bentham 和 Elsevier 数据库等多个不同的搜索引擎,进行文献综述,以研究治疗脑缺血的 HIF-1α 通路的药理调节:结果:各种信号通路,如丝裂原活化蛋白激酶(MAPK)、Janus 激酶/信号转导和激活因子(JAK/STAT)、磷脂酰肌醇-3-激酶(PI3-K)和 cAMP 反应元件结合蛋白(CREB)在调节 HIF-1α 通路中发挥着重要作用,有助于预防脑缺血的发病机制:结论:通过各种分子信号通路(如 PI3-K、MAPK、CREB 和 JAK/STAT 激活剂)对 HIF-1α 通路进行药理调节,为未来干预和治疗脑缺血提供了广阔的前景。
{"title":"Hypoxia-inducible Factor-1α Pathway in Cerebral Ischemia: From Molecular Mechanisms to Therapeutic Targets.","authors":"Veerta Sharma, Thakur Gurjeet Singh","doi":"10.2174/0118715273324551241008111827","DOIUrl":"https://doi.org/10.2174/0118715273324551241008111827","url":null,"abstract":"<p><strong>Introduction: </strong>Ischemic injury to the brain can result in a variety of life-threatening conditions, mortality, or varying degrees of disability. Hypoxia-inducible factor 1α (HIF 1α), an oxygen- sensitive transcription factor that controls the adaptive metabolic response to hypoxia, is a critical constituent of cerebral ischemia. It participates in numerous processes, such as metabolism, proliferation, and angiogenesis, and plays a major role in cerebral ischemia.</p><p><strong>Methods: </strong>Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the pharmacological modulation of HIF-1α pathways for the treatment of cerebral ischemia.</p><p><strong>Results: </strong>Various signalling pathways, such as Mitogen-activated protein kinase (MAPK), Janus kinase/ signal transducers and activators (JAK/STAT), Phosphoinositide-3-kinase (PI3-K), and cAMPresponse element binding protein (CREB) play a vital role in modulation of HIF-1α pathway, which helps in preventing the pathogenesis of cerebral ischemia.</p><p><strong>Conclusion: </strong>The pharmacological modulation of the HIF-1α pathway via various molecular signalling pathways, such as PI3-K, MAPK, CREB, and JAK/STAT activators, offer a promising prospect for future interventions and treatment for cerebral ischemia.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142483132","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Valproic Acid and Celecoxib Enhance the Effect of Temozolomide on Glioblastoma Cells. 丙戊酸和塞来昔布可增强替莫唑胺对胶质母细胞瘤细胞的作用
Pub Date : 2024-10-18 DOI: 10.2174/0118715273330268241008220702
Oleg Pak, Alexandra Kosianova, Sergei Zaitsev, Aruna Sharma, Hari Sharma, Igor Bryukhovetskiy

Introduction: Glioblastoma (GB) is one of the deadliest human brain tumors. The prognosis is unfavorable, chemotherapy with temozolomide (TMZ) may extend the survival period for a patient. The paper aims to evaluate the survival rates among relapsing GB patients, who have been treated with valproic acid (VPA), and to study its effect on tumor cells when combined with TMZ and celecoxib (CXB).

Materials and methods: The research is based on retrospective analysis of the data from GB patients who had been treated with VPA as a part of a complex treatment protocol and reoperated due to a GB relapse. The experimental study involved cancer cells of C6, U87, and T98G lines. GB was modeled on Wistar rats. The research was approved by the ethics committee. Differences in groups were considered significant at p < 0.05 Results: The median of overall survival among GB patients who took VPA was 22 months, and for those who did not take VPA - 13 months. The in vitro experiment showed the half-maximal inhibitory concentration (IC50) of TMZ for various lines of cancer cells (CCs) varying from 435.3 to 844 μM. IC50 VPA for CCs of U87MG, T98G, and С6 lines was 1510, 3900, and 3600 μM: IC50 CXB for those lines of CCs was 30.1 μM, 41.07, and 48.4 μM respectively. VPA significantly enhanced the anti-glioma effect of TMZ on the U87 line of CCs, while CCs of C6 and T98G lines proved to be most susceptible to the combination of CXB and TMZ. The combination of VPA with CXB increased the anti-glioma effect of TMZ both in vitro and in vivo, also reducing the tumor size (р < 0.05) and prolonging the survival period among experimental animals.

Conclusion: VPA and CXB enhance the effect of TMZ on glioblastoma cells.

简介胶质母细胞瘤(GB)是最致命的人类脑肿瘤之一。预后很差,替莫唑胺(TMZ)化疗可延长患者的生存期。本文旨在评估接受丙戊酸(VPA)治疗的复发性GB患者的生存率,并研究其与TMZ和塞来昔布(CXB)联合使用时对肿瘤细胞的影响:研究基于对曾接受过 VPA 治疗的 GB 患者数据的回顾性分析,VPA 是复杂治疗方案的一部分,患者因 GB 复发而再次接受手术。实验研究涉及 C6、U87 和 T98G 株系的癌细胞。GB以Wistar大鼠为模型。研究获得了伦理委员会的批准。组间差异以 P < 0.05 为显著结果:服用 VPA 的 GB 患者的总生存期中位数为 22 个月,未服用 VPA 的患者的总生存期中位数为 13 个月。体外实验显示,TMZ 对各种癌细胞(CCs)的半最大抑制浓度(IC50)从 435.3 μM 到 844 μM 不等。VPA 对 U87MG、T98G 和 С6 株系 CC 的 IC50 分别为 1510、3900 和 3600 μM:对这些株系 CC 的 IC50 CXB 分别为 30.1 μM、41.07 和 48.4 μM。VPA 能明显增强 TMZ 对 U87 株 CCs 的抗胶质瘤作用,而 C6 和 T98G 株 CCs 对 CXB 和 TMZ 的联合作用最敏感。VPA与CXB的联合用药在体外和体内都增强了TMZ的抗胶质瘤作用,同时还缩小了肿瘤体积(р < 0.05),延长了实验动物的生存期:结论:VPA和CXB能增强TMZ对胶质母细胞瘤细胞的作用。
{"title":"Valproic Acid and Celecoxib Enhance the Effect of Temozolomide on Glioblastoma Cells.","authors":"Oleg Pak, Alexandra Kosianova, Sergei Zaitsev, Aruna Sharma, Hari Sharma, Igor Bryukhovetskiy","doi":"10.2174/0118715273330268241008220702","DOIUrl":"https://doi.org/10.2174/0118715273330268241008220702","url":null,"abstract":"<p><strong>Introduction: </strong>Glioblastoma (GB) is one of the deadliest human brain tumors. The prognosis is unfavorable, chemotherapy with temozolomide (TMZ) may extend the survival period for a patient. The paper aims to evaluate the survival rates among relapsing GB patients, who have been treated with valproic acid (VPA), and to study its effect on tumor cells when combined with TMZ and celecoxib (CXB).</p><p><strong>Materials and methods: </strong>The research is based on retrospective analysis of the data from GB patients who had been treated with VPA as a part of a complex treatment protocol and reoperated due to a GB relapse. The experimental study involved cancer cells of C6, U87, and T98G lines. GB was modeled on Wistar rats. The research was approved by the ethics committee. Differences in groups were considered significant at p < 0.05 Results: The median of overall survival among GB patients who took VPA was 22 months, and for those who did not take VPA - 13 months. The in vitro experiment showed the half-maximal inhibitory concentration (IC50) of TMZ for various lines of cancer cells (CCs) varying from 435.3 to 844 μM. IC50 VPA for CCs of U87MG, T98G, and С6 lines was 1510, 3900, and 3600 μM: IC50 CXB for those lines of CCs was 30.1 μM, 41.07, and 48.4 μM respectively. VPA significantly enhanced the anti-glioma effect of TMZ on the U87 line of CCs, while CCs of C6 and T98G lines proved to be most susceptible to the combination of CXB and TMZ. The combination of VPA with CXB increased the anti-glioma effect of TMZ both in vitro and in vivo, also reducing the tumor size (р < 0.05) and prolonging the survival period among experimental animals.</p><p><strong>Conclusion: </strong>VPA and CXB enhance the effect of TMZ on glioblastoma cells.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142483134","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An Insight into Medicinal Chemistry and SAR Studies of Cholinesterase and BACE-1 Inhibitors for Alzheimer's Disease. 深入了解治疗阿尔茨海默病的胆碱酯酶和 BACE-1 抑制剂的药物化学和 SAR 研究。
Pub Date : 2024-10-16 DOI: 10.2174/0118715273315191241002115155
Abhimannu Shome, Keshav Taruneshwar Jha, Chahat, Viney Chawla, Pooja A Chawla

Alzheimer's Disease (AD) is a serious neurodegenerative condition that predominantly impacts the cholinergic neurons of the entorhinal cortex and hippocampal regions, playing a critical role in learning, navigation, and brain processing. This paper aims to discuss the three main hypotheses of Alzheimer's disease, focusing on neurotoxicity and neurodegeneration caused by mitochondrial dysfunction and ROS production, particularly analyzing the susceptibility differences between genders. Our comprehensive review focuses on significant findings from the past five years, particularly on Cholinesterase (ChE) and BACE-1 inhibitors. Researchers have conducted a detailed analysis of in vitro, in silico, and in vivo data, incorporating extensive Structure-Activity Relationship (SAR) studies. The reviewed papers have been sourced from platforms, such as Google Scholar, Semantic Scholar, and ClinicalTrials.gov, and have been selected based on their AChE and BACE-1 inhibitory activity and structural motif similarity. The review identifies the most effective compounds targeting ChE and BACE-1, highlighting acridine, dihydropyridine, and thiazole-coumarin hybrids for ChE inhibition, and oxadiazole, benzofuran, and dihydropyrimidinone for BACE-1 inhibition. This demonstrates a diverse array of potent heterocyclic hybrids. The review presents a varied compilation of scaffolds showing promise in treating Alzheimer's disease, highlighting the potential of specific compounds against ChE and BACE-1. Given the critical insights derived from our analysis, we posit that this compilation will substantially contribute to the ongoing efforts to combat neurodegeneration and prolong dementia, underscoring the importance of continuous research in this domain.

阿尔茨海默病(AD)是一种严重的神经退行性疾病,主要影响内叶皮层和海马区的胆碱能神经元,这些神经元在学习、导航和大脑处理过程中发挥着关键作用。本文旨在讨论阿尔茨海默病的三个主要假说,重点是线粒体功能障碍和 ROS 生成导致的神经毒性和神经变性,尤其是分析两性之间的易感性差异。我们的综合综述重点关注过去五年的重大发现,尤其是胆碱酯酶(ChE)和 BACE-1 抑制剂。研究人员对体外、硅学和体内数据进行了详细分析,并进行了广泛的结构-活性关系(SAR)研究。综述论文来自 Google Scholar、Semantic Scholar 和 ClinicalTrials.gov 等平台,根据其 AChE 和 BACE-1 抑制活性和结构相似性进行筛选。综述确定了针对胆碱酯酶和 BACE-1 的最有效化合物,重点介绍了抑制胆碱酯酶的吖啶、二氢吡啶和噻唑-香豆素杂交化合物,以及抑制 BACE-1 的噁二唑、苯并呋喃和二氢嘧啶酮。这展示了多种多样的强效杂环化合物。本综述汇集了各种有望治疗阿尔茨海默病的支架,突出了特定化合物对 ChE 和 BACE-1 的潜在作用。考虑到我们的分析所得出的重要见解,我们认为这篇综述将极大地推动目前对抗神经变性和延长痴呆症的工作,并强调了在这一领域持续开展研究的重要性。
{"title":"An Insight into Medicinal Chemistry and SAR Studies of Cholinesterase and BACE-1 Inhibitors for Alzheimer's Disease.","authors":"Abhimannu Shome, Keshav Taruneshwar Jha, Chahat, Viney Chawla, Pooja A Chawla","doi":"10.2174/0118715273315191241002115155","DOIUrl":"https://doi.org/10.2174/0118715273315191241002115155","url":null,"abstract":"<p><p>Alzheimer's Disease (AD) is a serious neurodegenerative condition that predominantly impacts the cholinergic neurons of the entorhinal cortex and hippocampal regions, playing a critical role in learning, navigation, and brain processing. This paper aims to discuss the three main hypotheses of Alzheimer's disease, focusing on neurotoxicity and neurodegeneration caused by mitochondrial dysfunction and ROS production, particularly analyzing the susceptibility differences between genders. Our comprehensive review focuses on significant findings from the past five years, particularly on Cholinesterase (ChE) and BACE-1 inhibitors. Researchers have conducted a detailed analysis of in vitro, in silico, and in vivo data, incorporating extensive Structure-Activity Relationship (SAR) studies. The reviewed papers have been sourced from platforms, such as Google Scholar, Semantic Scholar, and ClinicalTrials.gov, and have been selected based on their AChE and BACE-1 inhibitory activity and structural motif similarity. The review identifies the most effective compounds targeting ChE and BACE-1, highlighting acridine, dihydropyridine, and thiazole-coumarin hybrids for ChE inhibition, and oxadiazole, benzofuran, and dihydropyrimidinone for BACE-1 inhibition. This demonstrates a diverse array of potent heterocyclic hybrids. The review presents a varied compilation of scaffolds showing promise in treating Alzheimer's disease, highlighting the potential of specific compounds against ChE and BACE-1. Given the critical insights derived from our analysis, we posit that this compilation will substantially contribute to the ongoing efforts to combat neurodegeneration and prolong dementia, underscoring the importance of continuous research in this domain.</p>","PeriodicalId":93947,"journal":{"name":"CNS & neurological disorders drug targets","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142483130","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
CNS & neurological disorders drug targets
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1